UniGene: Hs.306721
Recombinant Human Putative transmembrane protein LOC100289255 is characterized by a molecular weight of approximately 8,713 Da as determined by mass spectrometry and SDS-PAGE analysis. The protein typically contains an N-terminal tag and may also include a C-terminal tag depending on the expression system utilized. The protein is available in both lyophilized and liquid formulations, with purity typically at or exceeding 85% as confirmed by SDS-PAGE analysis. Expression systems for this recombinant protein include E. coli, yeast, baculovirus, and mammalian cell platforms, each offering distinct post-translational modification profiles that may impact functional studies .
For optimal preparation and storage of LOC100289255, the following protocol is recommended: Store lyophilized protein at -20°C for routine storage or -80°C for extended preservation periods. When reconstituting lyophilized protein, use sterile buffer conditions appropriate for downstream applications (typically PBS or Tris-based buffers with physiological salt concentrations). Following reconstitution, prepare small working aliquots to avoid repeated freeze-thaw cycles, which can significantly compromise protein integrity. Working aliquots should be stored at 4°C and used within one week, while remaining stock should be returned to -20°C or -80°C. Prior to experimental use, briefly centrifuge the protein vial to collect any material that may have adhered to the cap during storage or shipping .
The selection of an expression system for LOC100289255 should be guided by the specific research objectives and required protein characteristics. E. coli systems offer high yield and cost-efficiency but lack eukaryotic post-translational modifications that may be critical for transmembrane protein functionality. Yeast expression systems (such as Pichia pastoris) provide moderate yields with basic eukaryotic modifications and may represent a balanced choice for initial characterization studies. Baculovirus expression systems offer more complete post-translational modifications and have demonstrated success with many transmembrane proteins similar to LOC100289255. Mammalian cell systems (typically HEK293 or CHO) provide the most physiologically relevant modifications but at higher production costs and potentially lower yields. Experimental validation comparing protein functionality across expression platforms is recommended for studies where native conformation and activity are critical parameters .
A comprehensive validation approach for LOC100289255 expression and purification should include multiple analytical methods. Begin with SDS-PAGE analysis under both reducing and non-reducing conditions to assess purity and potential oligomeric states, aiming for ≥85% purity. Western blotting using tag-specific antibodies (against N-terminal or C-terminal tags) provides confirmation of full-length protein expression. Mass spectrometry analysis (MALDI-TOF or ESI-MS) should be employed to verify the expected molecular weight of 8,713 Da and identify any post-translational modifications. For transmembrane proteins like LOC100289255, circular dichroism spectroscopy is valuable for secondary structure analysis, particularly to confirm proper folding of the protein's transmembrane regions. Additionally, size-exclusion chromatography can assess aggregation state and homogeneity. For functional validation, consider developing binding assays or activity tests based on predicted functions of transmembrane proteins, such as membrane integration assays using artificial liposomes .
For investigating protein-protein interactions of LOC100289255, a multi-technique approach is recommended. Co-immunoprecipitation (Co-IP) assays using antibodies against the protein's tags represent an accessible starting point, ideally conducted in both endogenous expression systems and with overexpressed constructs. For detailed binding kinetics, surface plasmon resonance (SPR) or biolayer interferometry (BLI) provide quantitative measurements of association and dissociation rates. Proximity ligation assays (PLA) offer visualization of interactions in situ with subcellular resolution. Yeast two-hybrid screening may identify novel interaction partners, though results should be confirmed with alternative methods due to potential false positives with transmembrane proteins. For high-confidence interactome mapping, quantitative proteomics approaches such as BioID or APEX proximity labeling, followed by mass spectrometry, are particularly valuable for transmembrane proteins where traditional interaction assays may be challenging. Based on known patterns of transmembrane protein function, investigating interactions with ER stress response proteins (similar to TMEM33's interaction with PERK) could be a productive direction for LOC100289255 research .
When investigating the subcellular localization of LOC100289255, employ multiple complementary approaches to ensure accurate characterization. Confocal microscopy with fluorescently tagged LOC100289255 constructs represents a primary approach, though tag position (N-terminal versus C-terminal) should be carefully considered as it may affect trafficking signals. Counterstaining with established organelle markers (particularly ER, Golgi, plasma membrane, and endosomal markers) is essential for precise localization determination. Subcellular fractionation followed by Western blotting provides biochemical verification of microscopy results and should include verification using marker proteins for each cellular compartment. Protease protection assays can determine membrane topology, revealing which protein domains face the cytosol versus the lumen. For transmembrane proteins like LOC100289255, analysis of the relative levels in detergent-resistant membrane fractions may provide insight into potential lipid raft association. Super-resolution microscopy techniques such as STORM or PALM are recommended for detailed analysis of transmembrane protein distribution within membrane subdomains .
Based on structural similarities with other transmembrane proteins, LOC100289255 may participate in endoplasmic reticulum (ER) stress response pathways. Methodologically, this hypothesis can be tested by monitoring changes in LOC100289255 expression levels during experimentally induced ER stress using qRT-PCR, Western blotting, and immunofluorescence. Researchers should expose cells to established ER stress inducers (tunicamycin, thapsigargin, DTT, or brefeldin A) and measure temporal expression changes of LOC100289255 relative to known ER stress markers such as BiP/GRP78, CHOP, and XBP1 splicing. To assess functional involvement, siRNA-mediated knockdown or CRISPR-Cas9 knockout of LOC100289255 followed by analysis of UPR signaling components (phosphorylation states of PERK, IRE1α, and ATF6 processing) would provide insight into its role in stress adaptation. Co-immunoprecipitation assays should be conducted to test for direct interactions with key UPR sensors, similar to the established interaction between TMEM33 and PERK. Particular attention should be paid to effects on the PERK-eIF2α-ATF4 and IRE1-XBP1 signaling axes, as these pathways are central to cellular adaptation to ER stress conditions .
To investigate potential prognostic significance of LOC100289255 in cancer contexts, researchers should implement a systematic analytical approach. Begin with comprehensive expression analysis across cancer tissue microarrays using immunohistochemistry with validated antibodies against LOC100289255, comparing expression in tumor versus matched normal tissues. This should be complemented by mining public cancer genomics databases (TCGA, ICGC) for LOC100289255 expression correlations with patient survival across multiple cancer types. For cellular mechanistic studies, stable overexpression and knockdown of LOC100289255 in relevant cancer cell lines should be established, followed by assessment of proliferation (MTT/MTS assays), migration (wound healing assays), invasion (transwell assays), and apoptotic resistance. The expression correlation between LOC100289255 and established biomarkers should be analyzed through both bioinformatic approaches and experimental validation. Kaplan-Meier survival analysis stratified by LOC100289255 expression levels will determine potential prognostic value, which should be confirmed through multivariate Cox regression analysis to establish if LOC100289255 expression represents an independent prognostic factor. This approach mirrors successful methodologies used for other prognostic markers such as linc00675 in pancreatic cancer research .
For comparative functional analysis between LOC100289255 and other transmembrane proteins, researchers should employ a structured approach combining bioinformatic prediction with experimental validation. Begin with comprehensive sequence alignment and structural modeling to identify conserved domains and motifs shared with well-characterized transmembrane proteins, particularly focusing on TMEM family proteins with established functions. Domain swap experiments, where specific regions of LOC100289255 are exchanged with corresponding regions from functionally characterized transmembrane proteins, can identify critical functional domains. Parallel knockdown/knockout experiments of LOC100289255 and related proteins followed by transcriptomic profiling will reveal shared and distinct regulatory networks. For transmembrane proteins involved in stress response pathways, compare induction kinetics of LOC100289255 with those of established stress-responsive transmembrane proteins under identical experimental conditions. Proteomic interactome mapping using standardized protocols across multiple transmembrane proteins will identify shared binding partners and potential functional complexes. These comparative approaches should be designed with appropriate controls and standardized experimental conditions to allow direct comparison across different protein family members .
Purification of functional transmembrane proteins like LOC100289255 presents several technical challenges that require specific methodological solutions. Protein aggregation during extraction and purification can be addressed by screening multiple detergents (starting with milder options like DDM, LMNG, or digitonin) and detergent-to-protein ratios to optimize solubilization conditions. Low expression yields common with transmembrane proteins can be improved by utilizing specialized expression strains (such as C41/C43 for E. coli) or codon-optimization of the construct for the chosen expression system. Improper folding may be mitigated by expression at reduced temperatures (16-25°C) and co-expression with molecular chaperones. For obtaining protein with native conformation, consider nanodiscs or liposome reconstitution following purification. Proteolytic degradation during purification should be addressed by including a comprehensive protease inhibitor cocktail throughout all purification steps and minimizing processing time. If conventional immobilized metal affinity chromatography (IMAC) yields insufficiently pure protein, implement a multi-step purification strategy incorporating ion exchange chromatography and size exclusion chromatography. Functional validation after purification is essential and should include activity assays specific to predicted functions of LOC100289255 .
Developing reliable detection methods for endogenous LOC100289255 requires a comprehensive validation approach. For antibody-based detection, commission custom antibodies against multiple epitopes of LOC100289255, preferably targeting both N-terminal and C-terminal regions. Validate antibody specificity through multiple approaches: Western blotting comparing wild-type cells with CRISPR knockout/knockdown cells, immunoprecipitation followed by mass spectrometry, and peptide competition assays. For transcript detection, design and validate qRT-PCR primers spanning exon-exon junctions to prevent genomic DNA amplification, with efficiency testing using standard curves and melt curve analysis to confirm single-product amplification. RNA-seq data should be analyzed with specific attention to read coverage across all exons to identify potential splice variants. For in situ detection, validate immunohistochemistry protocols across multiple tissue types with appropriate positive and negative controls, including peptide blocking and CRISPR knockout tissues. When investigating protein expression in disease contexts, use paired normal-pathological samples from the same patient to control for individual variation. All detection methods should be cross-validated using at least two independent techniques to ensure reliable quantification of endogenous LOC100289255 .
For comprehensive structure-function characterization of LOC100289255, implement advanced analytical techniques targeted to transmembrane protein analysis. Cryo-electron microscopy represents a powerful approach for structural determination without the need for protein crystallization, which is often challenging for transmembrane proteins. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) can map dynamic conformational changes and identify regions involved in protein-protein interactions or ligand binding. Site-directed mutagenesis of predicted functional residues followed by activity assays will establish critical structure-function relationships. For transmembrane topology analysis, cysteine scanning mutagenesis combined with accessibility assays provides detailed mapping of membrane-spanning regions. Molecular dynamics simulations based on homology models can predict protein behavior in membrane environments and guide experimental design. For functional domains, produce truncated constructs systematically removing specific domains and assess changes in localization, interaction partners, and cellular responses. Advanced live-cell imaging techniques such as Förster resonance energy transfer (FRET) or fluorescence recovery after photobleaching (FRAP) are valuable for studying dynamic behavior of LOC100289255 in living cells, particularly for monitoring potential conformational changes in response to cellular stress conditions .